Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
2.
Signal Transduct Target Ther ; 5(1): 14, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32296036

ABSTRACT

Sirtuin 3 (SIRT3) is a deacetylase that modulates proteins that control metabolism and protects against oxidative stress. Modulation of SIRT3 activity has been proposed as a promising therapeutic target for ameliorating metabolic diseases and associated cardiac disturbances. In this study, we investigated the role of SIRT3 in inflammation and fibrosis in the heart using male mice with constitutive and systemic deletion of SIRT3 and human cardiac AC16 cells. SIRT3 knockout mice showed cardiac fibrosis and inflammation that was characterized by augmented transcriptional activity of AP-1. Consistent with this, SIRT3 overexpression in human and neonatal rat cardiomyocytes partially prevented the inflammatory and profibrotic response induced by TNF-α. Notably, these effects were associated with a decrease in the mRNA and protein levels of FOS and the DNA-binding activity of AP-1. Finally, we demonstrated that SIRT3 inhibits FOS transcription through specific histone H3 lysine K27 deacetylation at its promoter. These findings highlight an important function of SIRT3 in mediating the often intricate profibrotic and proinflammatory responses of cardiac cells through the modulation of the FOS/AP-1 pathway. Since fibrosis and inflammation are crucial in the progression of cardiac hypertrophy, heart failure, and diabetic cardiomyopathy, our results point to SIRT3 as a potential target for treating these diseases.


Subject(s)
Fibrosis/genetics , Heart Failure/genetics , Proto-Oncogene Proteins c-fos/genetics , Sirtuin 3/genetics , Transcription Factor AP-1/genetics , Animals , Fibrosis/pathology , Heart , Heart Failure/pathology , Histones/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/genetics , Protein Processing, Post-Translational/genetics , Rats
3.
Mutat Res ; 806: 88-97, 2017 12.
Article in English | MEDLINE | ID: mdl-28283188

ABSTRACT

The effect of ionizing irradiation on cytoplasmic organelles is often underestimated because the general dogma considers direct DNA damage in the nuclei to be the primary cause of radiation induced toxicity. Using a precision microbeam irradiator, we examined the changes in mitochondrial dynamics and functions triggered by targeted cytoplasmic irradiation with α-particles. Mitochondrial dysfunction induced by targeted cytoplasmic irradiation led to activation of autophagy, which degraded dysfunctional mitochondria in order to maintain cellular energy homeostasis. The activation of autophagy was cytoplasmic irradiation-specific and was not detected in nuclear irradiated cells. This autophagic process was oxyradical-dependent and required the activity of the mitochondrial fission protein dynamin related protein 1 (DRP1). The resultant mitochondrial fission induced phosphorylation of AMP activated protein kinase (AMPK) which leads to further activation of the extracellular signal-related kinase (ERK) 1/2 with concomitant inhibition of the mammalian target of rapamycin (mTOR) to initiate autophagy. Inhibition of autophagy resulted in delayed DNA damage repair and decreased cell viability, which supports the cytoprotective function of autophagy. Our results reveal a novel mechanism in which dysfunctional mitochondria are degraded by autophagy in an attempt to protect cells from toxic effects of targeted cytoplasmic radiation.


Subject(s)
Alpha Particles , Apoptosis/radiation effects , Autophagy/radiation effects , Cytoplasm/radiation effects , Epithelial Cells/pathology , Respiratory System/pathology , Cells, Cultured , Epithelial Cells/radiation effects , Humans , Mitochondrial Dynamics , Reactive Oxygen Species/metabolism , Respiratory System/radiation effects , TOR Serine-Threonine Kinases/metabolism
4.
Basic Res Cardiol ; 112(1): 1, 2017 01.
Article in English | MEDLINE | ID: mdl-27837311

ABSTRACT

Chronic increased workload of the human heart causes ventricular hypertrophy, re-expression of the atrial essential myosin light chain (hALC-1), and improved contractile function. Although hALC-1 is an important positive inotropic regulator of the human heart, little is known about its regulation. Therefore, we investigated the role of the sex hormone 17ß-estradiol (E2) on hALC-1 gene expression, the underlying molecular mechanisms, and the impact of this regulatory process on cardiac contractile function. We showed that E2 attenuated hALC-1 expression in human atrial tissues of both sexes and in human ventricular AC16 cells. E2 induced the nuclear translocation of estrogen receptor alpha (ERα) and hALC-1 in AC16 cells, where they cooperatively regulate the transcriptional activity of hALC-1 gene promoter. E2-activated ERα required the estrogen response element (ERE) motif within the hALC-1 gene promoter to reduce its transcriptional activity (vehicle: 15.55 ± 4.80 vs. E2: 6.51 ± 3.69; ~2 fold). This inhibitory effect was potentiated in the presence of hALC-1 (vehicle: 11.13 ± 3.66 vs. E2: 2.18 ± 1.10; ~5 fold), and thus, hALC-1 acts as a co-repressor of ERα-mediated transcription. Yeast two-hybrid screening of a human heart cDNA library revealed that ERα interacts physically with hALC-1 in the presence of E2. This interaction was confirmed by Co-Immunoprecipitation and immunofluorescence in human atrium. As a further novel effect, we showed that chronic E2-treatment of adult mouse cardiomyocytes overexpressing hALC-1 resulted in reduced cell-shortening amplitude and twitching kinetics of these cells independent of Ca2+ activation levels. Together, our data showed that the expression of hALC-1 gene is, at least partly, regulated by E2/ERα, while hALC-1 acts as a co-repressor. The inotropic effect of hALC-1 overexpression in cardiomyocytes can be significantly repressed by E2.


Subject(s)
Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation/genetics , Myocardial Contraction/physiology , Myosin Light Chains/biosynthesis , Animals , Blotting, Western , Female , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Myocytes, Cardiac/metabolism , Myosin Light Chains/genetics , Polymerase Chain Reaction , Two-Hybrid System Techniques
5.
Dis Model Mech ; 8(9): 1081-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26112171

ABSTRACT

miR-146a is a microRNA whose transcript levels are induced in the heart upon activation of NF-κB, a transcription factor induced by pro-inflammatory molecules (such as TNF-α) that is strongly related to the pathogenesis of cardiac disorders. The main goal of this study consisted of studying new roles of miR-146a in cardiac pathological processes caused by the pro-inflammatory cytokine TNF-α. Our results demonstrate that miR-146a transcript levels were sharply increased in cardiac ventricular tissue of transgenic mice with specific overexpression of TNF-α in the heart, and also in a cardiomyocyte cell line of human origin (AC16) exposed to TNF-α. Among all the in silico predicted miR-146a target genes, Fos mRNA and protein levels notably decreased after TNF-α treatment or miR-146a overexpression. These changes correlated with a diminution in the DNA-binding activity of AP-1, the Fos-containing transcription factor complex. Interestingly, AP-1 inhibition was accompanied by a reduction in matrix metalloproteinase (MMP)-9 mRNA levels in human cardiac cells. The specific regulation of this MMP by miR-146a was further confirmed at the secretion and enzymatic activity levels, as well as after anti-miR-mediated miR-146a inhibition. The results reported here demonstrate that Fos is a direct target of miR-146a activity and that downregulation of the Fos-AP-1 pathway by miR-146a has the capacity to inhibit MMP-9 activity. Given that MMP-9 is an AP-1 target gene involved in cardiac remodeling, myocardial dysfunction and progression of heart failure, these findings suggest that miR-146a might be a new and promising therapeutic tool for treating cardiac disorders associated with enhanced inflammation in the heart.


Subject(s)
Gene Expression Regulation , MicroRNAs/physiology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Line , Humans , Immune System , Inflammation , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Arch Toxicol ; 89(7): 1057-70, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24961358

ABSTRACT

Although kidney is a target organ of arsenic cytotoxicity, the underlying mechanisms of arsenic-induced nephrotoxicity remain poorly understood. As tetramethylpyrazine (TMP) has recently been found to be a renal protectant in multiple kidney injuries, we hypothesize that TMP could suppress arsenic nephrotoxicity. In this study, human renal proximal tubular epithelial cell line HK-2 was used to elucidate the precise mechanisms of arsenic nephrotoxicity as well as the protective mechanism of TMP in these cells. Sodium arsenite exposure dramatically increased cellular reactive oxygen species (ROS) production, decreased levels of cellular glutathione (GSH), decreased cytochrome c oxidase activity and mitochondrial membrane potential, which indicated mitochondrial dysfunction. On the other hand, sodium arsenite activated pro-inflammatory signals, including ß-catenin, nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (MAPK), tumor necrosis factor alpha and cyclooxygenase-2 (COX-2). Small molecule inhibitors of NF-κB and p38 MAPK blocked arsenic-induced COX-2 expression, suggesting arsenic-induced COX-2 up-regulation was NF-κB- and p38 MAPK-dependent. Finally, sodium arsenite induced autophagy in HK-2 cells at early phase (6 h) and the subsequent apoptosis at 24 h. Treatment by TMP or by the antioxidant N-acetylcysteine decreased arsenic-induced ROS production, enhanced GSH levels, prevented mitochondria dysfunction and suppressed the activation of pro-inflammatory signals and the development of autophagy and apoptosis. Our results suggested that TMP may be used as a new potential therapeutic agent to prevent arsenic-induced nephrotoxicity by suppressing these pathological processes.


Subject(s)
Apoptosis/drug effects , Arsenites/toxicity , Autophagy/drug effects , Inflammation Mediators/metabolism , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/chemistry , Mitochondria/drug effects , Oxidative Stress/drug effects , Pyrazines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sodium Compounds/toxicity , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Cell Line , Cytoprotection , Dose-Response Relationship, Drug , Glutathione/metabolism , Humans , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Time Factors
7.
Clín. investig. arterioscler. (Ed. impr.) ; 26(6): 255-267, nov.-dic. 2014. ilus, tab
Article in Spanish | IBECS | ID: ibc-129674

ABSTRACT

Introducción El estrés del retículo endoplasmático (RE) se ha relacionado con distintas enfermedades cardiovasculares, como la arteriosclerosis y la hipertrofia e insuficiencia cardíacas. Este estrés del RE altera la señalización de la insulina, contribuyendo al desarrollo de la resistencia a la insulina y la diabetes. Diversos estudios han demostrado que PPARalfa inhibe el estrés del RE, por lo que el objetivo de este trabajo consistió en investigar si la activación de este receptor nuclear era capaz de prevenir el estrés del RE inducido por ácidos grasos saturados en células cardíacas, así como los mecanismos implicados. Métodos: Cardiomiocitos humanos AC16 fueron tratados con palmitato en presencia de diferentes activadores e inhibidores de AMPK y PPARalfa. Para los estudios in vivo, ratones macho fueron alimentados con una dieta rica en grasa (HFD). Posteriormente, se determinó la presencia de distintos marcadores de estrés del RE en células cardíacas por medio del análisis de la expresión génica y la acumulación proteica. Resultados: El palmitato y la dieta HFD indujeron el estrés del RE en células cardíacas, pues incrementaron diversos marcadores de este, como son la expresión génica de ATF3, BiP/GRP78 y CHOP, el splicing de XBP1 y la fosforilación de IRE-1α y eIF2alfa. El tratamiento con Wy-14,643, un agonista de PPARalfa, previno el incremento del estrés del RE inducido por palmitato por medio de la activación de la AMPK. Conclusión: Wy-14,643 podría ser útil para prevenir el estrés del RE y las enfermedades cardiovasculares asociadas en pacientes obesos, e incluso durante la cardiomiopatía diabética, por medio de la activación de AMPK


Introduction Endoplasmic reticulum (ER) stress has been linked to several cardiovascular diseases, such as atherosclerosis, heart failure and cardiac hypertrophy. ER stress impairs insulin signalling, thus contributing to the development of insulin resistance and diabetes. Since several studies have reported that PPARalfa may inhibit ER stress, the main aim of this study consisted in investigating whether activation of this nuclear receptor is able to prevent lipid-induced ER stress in cardiac cells, as well as studying the mechanisms involved. Methods: A cardiomyocyte cell line of human origin, AC16, was treated with palmitate in the presence or absence of several AMPK and PPARα pharmacological agonists and antagonists. For the in vivo studies, wild-type male mice were fed a standard diet, or a high-fat diet (HFD), for two months. At the end of the experiments, several ER stress markers were assessed in cardiac cells or in the mice hearts, using real-time RT-PCR and Western-blot analyses. Results: The results demonstrate that both palmitate and the HFD induced ER stress in cardiac cells, since they upregulated the expression (ATF3, BiP/GRP78 and CHOP), splicing (sXBP1), and phosphorylation (IRE-1α and eIF2α) of several ER stress markers. Interestingly, treatment with the PPARalfa agonist Wy-14,643 prevented an increase in the majority of these ER stress markers in human cardiac cells by means of AMPK activation. Conclusion: These data indicate that PPARα activation by Wy-14,643 might be useful to prevent the harmful effects of ER stress and associated cardiovascular diseases in obese patients, and even during diabetic cardiomyopathy, by enhancing AMPK activity


Subject(s)
Animals , Mice , Endoplasmic Reticulum Stress , Peroxisome Proliferator-Activated Receptors/pharmacokinetics , Myocytes, Cardiac , Atherosclerosis/physiopathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , /analysis , Protective Agents/pharmacokinetics
8.
Life Sci Space Res (Amst) ; 1: 80-88, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-25072018

ABSTRACT

High linear energy transfer (LET) radiation including α particles and heavy ions is the major type of radiation find in space and is considered a potential health risk for astronauts. Even though the chance that these high LET particles traversing through the cytoplasm of cells is higher than that through the nuclei, the contribution of targeted cytoplasmic irradiation, to the induction of genomic instability and other chromosomal damages induced by high LET radiation is not known. In the present study, we investigated whether mitochondria are the potential cytoplasmic target of high LET radiation in mediating cellular damage using a mitochondrial DNA (mtDNA) depleted (ρ0) human small airway epithelial (SAE) cell model and a precision charged particle microbeam with a beam width of merely one micron. Targeted cytoplasmic irradiation by high LET α particles induced DNA oxidative damage and double strand breaks in wild type ρ+ SAE cells. Furthermore, there was a significant increase in autophagy, micronuclei, which is an indication of genomic instability, together with the activation of nuclear factor kappa-B (NF-κB) and mitochondrial inducible nitric oxide synthase (iNOS) signaling pathways in ρ+ SAE cells. In contrast, ρ0 SAE cells exhibited a significantly lower response to these same endpoints examined after cytoplasmic irradiation with high LET α particles. The results indicate that mitochondria are essential in mediating cytoplasmic radiation induced genotoxic damage in mammalian cells. Furthermore, the findings may shed some light in the design of countermeasures for space radiation.

9.
Clin Investig Arterioscler ; 26(6): 255-67, 2014.
Article in Spanish | MEDLINE | ID: mdl-24709376

ABSTRACT

INTRODUCTION: Endoplasmic reticulum (ER) stress has been linked to several cardiovascular diseases, such as atherosclerosis, heart failure and cardiac hypertrophy. ER stress impairs insulin signalling, thus contributing to the development of insulin resistance and diabetes. Since several studies have reported that PPARα may inhibit ER stress, the main aim of this study consisted in investigating whether activation of this nuclear receptor is able to prevent lipid-induced ER stress in cardiac cells, as well as studying the mechanisms involved. METHODS: A cardiomyocyte cell line of human origin, AC16, was treated with palmitate in the presence or absence of several AMPK and PPARα pharmacological agonists and antagonists. For the in vivo studies, wild-type male mice were fed a standard diet, or a high-fat diet (HFD), for two months. At the end of the experiments, several ER stress markers were assessed in cardiac cells or in the mice hearts, using real-time RT-PCR and Western-blot analyses. RESULTS: The results demonstrate that both palmitate and the HFD induced ER stress in cardiac cells, since they upregulated the expression (ATF3, BiP/GRP78 and CHOP), splicing (sXBP1), and phosphorylation (IRE-1α and eIF2α) of several ER stress markers. Interestingly, treatment with the PPARα agonist Wy-14,643 prevented an increase in the majority of these ER stress markers in human cardiac cells by means of AMPK activation. CONCLUSION: These data indicate that PPARα activation by Wy-14,643 might be useful to prevent the harmful effects of ER stress and associated cardiovascular diseases in obese patients, and even during diabetic cardiomyopathy, by enhancing AMPK activity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/physiology , Myocytes, Cardiac/pathology , PPAR alpha/metabolism , Animals , Blotting, Western , Cell Line , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , PPAR alpha/agonists , Palmitates/administration & dosage , Pyrimidines/pharmacology , Real-Time Polymerase Chain Reaction
10.
Int J Cardiol ; 174(1): 110-8, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24767130

ABSTRACT

BACKGROUND: Chronic endoplasmic reticulum (ER) stress contributes to the apoptotic cell death in the myocardium, thereby playing a critical role in the development of cardiomyopathy. ER stress has been reported to be induced after high-fat diet feeding in mice and also after saturated fatty acid treatment in vitro. Therefore, since several studies have shown that peroxisome proliferator-activated receptor (PPAR)ß/δ inhibits ER stress, the main goal of this study consisted in investigating whether activation of this nuclear receptor was able to prevent lipid-induced ER stress in cardiac cells. METHODS AND RESULTS: Wild-type and transgenic mice with reduced PPARß/δ expression were fed a standard diet or a high-fat diet for two months. For in vitro studies, a cardiomyocyte cell line of human origin, AC16, was treated with palmitate and the PPARß/δ agonist GW501516. Our results demonstrate that palmitate induced ER stress in AC16 cells, a fact which was prevented after PPARß/δ activation with GW501516. Interestingly, the effect of GW501516 on ER stress occurred in an AMPK-independent manner. The most striking result of this study is that GW501516 treatment also upregulated the protein levels of beclin 1 and LC3II, two well-known markers of autophagy. In accordance with this, feeding on a high-fat diet or suppression of PPARß/δ in knockout mice induced ER stress in the heart. Moreover, PPARß/δ knockout mice also displayed a reduction in autophagic markers. CONCLUSION: Our data indicate that PPARß/δ activation might be useful to prevent the harmful effects of ER stress induced by saturated fatty acids in the heart by inducing autophagy.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/drug effects , PPAR delta/pharmacology , PPAR-beta/pharmacology , Palmitates/pharmacology , Animals , Cells, Cultured , Humans , Male , Mice , Mice, Knockout , Thiazoles/pharmacology
11.
Cardiovasc Res ; 102(3): 418-28, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24654233

ABSTRACT

AIMS: Oestrogen receptor alpha (ERα) and beta (ERß) are involved in the regulation of pathological myocardial hypertrophy (MH). We hypothesize that both ER are also involved in physiological MH. Therefore, we investigated the role of ER in exercise-induced physiological MH in loss-of-function models and studied potential mechanisms of action. METHODS AND RESULTS: We performed 1 and 8 weeks of voluntary cage wheel running (VCR) with male and female C57BL/6J wild-type (WT), ERα- and ERß-deleted mice. In line with other studies, female WT mice ran more than males (P ≤ 0.001). After 8 weeks of VCR, both sexes showed an increase in left ventricular mass (females: P ≤ 0.01 and males: P ≤ 0.05) with more pronounced MH in females (P < 0.05). As previously shown, female ERα-deleted mice run less than female WT mice (P ≤ 0.001). ERß-deleted mice showed similar running performance as WT mice (females vs. male: P ≤ 0.001), but did not develop MH. Only female WT mice showed an increase in phosphorylation of serine/threonine kinase (AKT), ERK1/2, p38-mitogen-activated protein kinase (MAPK), and ribosomal protein s6, as well as an increase in the expression of key regulators of mitochondrial function and mitochondrial respiratory chain proteins (complexes I, III, and V) after VCR. However, ERß deletion abolished all observed sex differences. Mitochondrial remodelling occurred in female WT-VCR mice, but not in female ERß-deleted mice. CONCLUSION: The sex-specific response of the heart to exercise is modulated by ERß. The greater increase in physiological MH in females is mediated by induction of AKT signalling, MAPK pathways, protein synthesis, and mitochondrial adaptation via ERß.


Subject(s)
Cardiomegaly/etiology , Estrogen Receptor beta/physiology , Physical Conditioning, Animal , Adaptation, Physiological , Animals , Cells, Cultured , Female , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/physiology , Oxidative Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , Receptors, Estrogen/physiology , Sex Characteristics , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cancer Res ; 73(22): 6700-10, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24080278

ABSTRACT

Direct DNA damage is often considered the primary cause of cancer in patients exposed to ionizing radiation or environmental carcinogens. Although mitochondria are known to play an important role in radiation-induced cellular response, the mechanisms by which cytoplasmic stimuli modulate mitochondrial dynamics and functions are largely unknown. In the present study, we examined changes in mitochondrial dynamics and functions triggered by α particle damage to the mitochondria in human small airway epithelial cells, using a precision microbeam irradiator with a beam width of 1 µm. Targeted cytoplasmic irradiation using this device resulted in mitochondrial fragmentation and a reduction of cytochrome c oxidase and succinate dehydrogenase activity, when compared with nonirradiated controls, suggesting a reduction in respiratory chain function. In addition, mitochondrial fragmentation or fission was associated with increased expression of the dynamin-like protein DRP1, which promotes mitochondrial fission. DRP1 inhibition by the drug mdivi-1 prevented radiation-induced mitochondrial fission, but respiratory chain function in mitochondria inhibited by radiation persisted for 12 hours. Irradiated cells also showed an increase in mitochondria-derived superoxide that could be quenched by dimethyl sulfoxide. Taken together, our results provide a mechanistic explanation for the extranuclear, nontargeted effects of ionizing radiation.


Subject(s)
Cytoplasm/radiation effects , GTP Phosphohydrolases/physiology , Microtubule-Associated Proteins/physiology , Mitochondria/radiation effects , Mitochondrial Dynamics/radiation effects , Mitochondrial Proteins/physiology , Apoptosis/drug effects , Cells, Cultured , DNA Damage/drug effects , Dynamins , GTP Phosphohydrolases/antagonists & inhibitors , Gene Expression/radiation effects , HCT116 Cells , Humans , Microtubule-Associated Proteins/antagonists & inhibitors , Mitochondria/physiology , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Quinazolinones/pharmacology , Reactive Oxygen Species/metabolism
13.
Int J Cardiol ; 167(6): 2507-16, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-22748497

ABSTRACT

BACKGROUND: Resveratrol is a grape polyphenol that prevents cardiac hypertrophy and protects the heart from ischemic injury, metabolic dysregulation, and inflammatory processes in several murine models. METHODS AND RESULTS: The aim of this study was to investigate the effects of resveratrol on the inflammatory processes in human cardiac AC16 cells in order to gain a better understanding of its cardioprotective mechanisms in the human heart. Resveratrol induced the DNA-binding activity of the pro-inflammatory transcription factor NF-κB in AC16 cells, and exacerbated the increase caused by tumor necrosis factor-α (TNF-α). In accordance with this, resveratrol increased the expression of the pro-inflammatory genes ICAM-1 (intercellular adhesion molecule-1) and TNF-α. In contrast, resveratrol decreased the expression of pro-inflammatory genes IL-6 (interleukin-6) and MCP-1 (monocyte chemoattractant protein-1). Likewise, resveratrol also induced inflammation in rat neonatal cardiomyocytes, and in the heart of mice fed a standard chow diet supplemented with resveratrol (1g/kg diet) for four months. Western-blot analyses revealed that NF-κB p65 subunit levels were upregulated in an IκB-dependent manner in the nuclei of resveratrol-treated human cardiac cells. Finally, resveratrol activated the signal transducer and activator of transcription 3 (STAT3) signaling and induced the expression of its anti-apoptotic downstream effector Bcl-xL, both involved in the cardioprotective survival activating factor enhancement (SAFE) pathway. CONCLUSIONS: Resveratrol enhanced NF-κB activity in human and murine cardiac cells, in a process that coincided with the activation of STAT3 and anti-apoptotic downstream effectors. Therefore, activation of the SAFE pathway by resveratrol might be involved in the cardioprotective effects of this compound.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Stilbenes/pharmacology , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Resveratrol
14.
Cardiovasc Res ; 96(3): 411-21, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22962310

ABSTRACT

AIMS: 17ß-Oestradiol (E2) and its receptors (ERα and ERß) are important regulators of physiological and pathological processes in the cardiovascular system. ER act in concert with other regulatory factors mediating oestrogenic effects. However, the underlying mechanisms modulating ER transcriptional activity are not fully elucidated. To gain better understanding of E2-induced ERα action in the human heart, we aimed to identify and functionally analyse interaction partners of ERα. METHODS AND RESULTS: Using yeast two-hybrid assays with a human heart cDNA library, we identified atrial natriuretic peptide precursor A (NPPA), a well-known cardiac hypertrophy marker, as a novel ERα interaction partner interacting in an E2-dependent manner. Mutation analyses and immunofluorescence data indicated that the LXXLL motif within NPPA is necessary for its E2-induced interaction with ERα, its action as a co-repressor of ERα, and its translocation into the nucleus of human and rat cardiomyocytes. Expression analysis and chromatin immunoprecipitation assays in a human left ventricular cardiomyocyte cell line, AC16, showed that NPPA interacts with E2/ERα, suppressing the transcriptional activity of ERα on E2-target genes, such as NPPA, connexin43, αactinin-2, nuclear factor of activated T-cells, and collagens I and III. CONCLUSION: We characterize for the first time an E2-regulated interaction of NPPA with ERα in cardiomyocytes, that may be crucial in physiological and/or pathological cardiac processes, thereby representing a potential therapeutic target.


Subject(s)
Atrial Natriuretic Factor/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Myocytes, Cardiac/drug effects , Active Transport, Cell Nucleus , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Cell Line , Chromatin Immunoprecipitation , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Myocytes, Cardiac/metabolism , Natriuretic Peptide, C-Type/genetics , Natriuretic Peptide, C-Type/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription, Genetic , Transfection , Two-Hybrid System Techniques
15.
Clín. investig. arterioscler. (Ed. impr.) ; 24(3): 131-140, mayo-jun. 2012. ilus, tab
Article in Spanish | IBECS | ID: ibc-105086

ABSTRACT

Introducción El consumo de dietas ricas en grasas se relaciona con alteraciones cardíacas caracterizadas por un proceso inflamatorio de baja intensidad mediado por NF-κB. PPARbeta/delta ha sido propuesto como potencial diana terapéutica para paliar el proceso inflamatorio asociado a alteraciones cardiovasculares. Sin embargo, se desconoce la implicación de este receptor en la respuesta inflamatoria inducida por lípidos en el corazón (..) (AU)


Introduction High-fat diet intake is associated with cardiac disorders characterised by a low-grade inflammatory process which involves NF-κB activation. PPARbeta/delta has been proposed as a potential therapeutic target to mitigate the inflammatory process related to cardiovascular disorders. However, the involvement of this receptor in lipid-induced inflammatory response in the heart is not yet known (..) (AU)


Subject(s)
Animals , Mice , PPAR-beta/pharmacokinetics , Inflammation/physiopathology , Inflammation Mediators/analysis , Lipids/adverse effects , NF-kappa B/physiology , Cytokines , Chemokines , Palmitates/pharmacokinetics , Mice, Knockout
16.
Orphanet J Rare Dis ; 7: 21, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22515166

ABSTRACT

BACKGROUND: Mutations in SCO2 cause cytochrome c oxidase deficiency (COX) and a fatal infantile cardioencephalomyopathy. SCO2 encodes a protein involved in COX copper metabolism; supplementation with copper salts rescues the defect in patients' cells. Bezafibrate (BZF), an approved hypolipidemic agent, ameliorates the COX deficiency in mice with mutations in COX10, another COX-assembly gene. METHODS: We have investigated the effect of BZF and copper in cells with SCO2 mutations using spectrophotometric methods to analyse respiratory chain activities and a luciferase assay to measure ATP production.. RESULTS: Individual mitochondrial enzymes displayed different responses to BZF. COX activity increased by about 40% above basal levels (both in controls and patients), with SCO2 cells reaching 75-80% COX activity compared to untreated controls. The increase in COX was paralleled by an increase in ATP production. The effect was dose-dependent: it was negligible with 100 µM BZF, and peaked at 400 µM BZF. Higher BZF concentrations were associated with a relative decline of COX activity, indicating that the therapeutic range of this drug is very narrow. Combined treatment with 100 µM CuCl2 and 200 µM BZF (which are only marginally effective when administered individually) achieved complete rescue of COX activity in SCO2 cells. CONCLUSIONS: These data are crucial to design therapeutic trials for this otherwise fatal disorder. The additive effect of copper and BZF will allow to employ lower doses of each drug and to reduce their potential toxic effects. The exact mechanism of action of BZF remains to be determined.


Subject(s)
Bezafibrate/pharmacology , Carrier Proteins/genetics , Copper/pharmacology , Cytochrome-c Oxidase Deficiency/genetics , Fibroblasts/drug effects , Mitochondrial Proteins/genetics , Mutation , Adenosine Triphosphate/metabolism , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Cytochrome-c Oxidase Deficiency/drug therapy , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Mitochondrial Proteins/metabolism , Molecular Chaperones
17.
J Biol Chem ; 287(22): 18429-39, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22493506

ABSTRACT

The role of serine palmitoyltransferase (SPT) and de novo ceramide biosynthesis in cardiac ceramide and sphingomyelin metabolism is unclear. To determine whether the de novo synthetic pathways, rather than ceramide uptake from circulating lipoproteins, is important for heart ceramide levels, we created cardiomyocyte-specific deficiency of Sptlc2, a subunit of SPT. Heart-specific Sptlc2-deficient (hSptlc2 KO) mice had a >35% reduction in ceramide, which was limited to C18:0 and very long chain ceramides. Sphingomyelinase expression, and levels of sphingomyelin and diacylglycerol were unchanged. But surprisingly phospholipids and acyl CoAs contained increased saturated long chain fatty acids. hSptlc2 KO mice had decreased fractional shortening and thinning of the cardiac wall. While the genes regulating glucose and fatty acid metabolism were not changed, expression of cardiac failure markers and the genes involved in the formation of extracellular matrices were up-regulated in hSptlc2 KO hearts. In addition, ER-stress markers were up-regulated leading to increased apoptosis. These results suggest that Sptlc2-mediated de novo ceramide synthesis is an essential source of C18:0 and very long chain, but not of shorter chain, ceramides in the heart. Changes in heart lipids other than ceramide levels lead to cardiac toxicity.


Subject(s)
Ceramides/metabolism , Heart/physiopathology , Myocardium/enzymology , Serine C-Palmitoyltransferase/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Cells, Cultured , In Situ Nick-End Labeling , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Serine C-Palmitoyltransferase/genetics
18.
Environ Health Perspect ; 120(6): 840-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22398240

ABSTRACT

BACKGROUND: The incidence of asbestos-induced human cancers is increasing worldwide, and considerable evidence suggests that reactive oxygen species (ROS) are important mediators of these diseases. Our previous studies suggested that mitochondria might be involved in the initiation of oxidative stress in asbestos-exposed mammalian cells. OBJECTIVE: We investigated whether mitochondria are a potential cytoplasmic target of asbestos using a mitochondrial DNA-depleted (ρ(0)) human small airway epithelial (SAE) cell model: ρ(0) SAE cells lack the capacity to produce mitochondrial ROS. METHODS: We examined nuclear DNA damage, micronuclei (MN), intracellular ROS production, and the expression of inflammation-related nuclear genes in both parental and ρ(0) SAE cells in response to asbestos treatment. RESULTS: Asbestos induced a dose-dependent increase in nuclear DNA oxidative damage and MN in SAE cells. Furthermore, there was a significant increase in intracellular oxidant production and activation of genes involved in nuclear factor κB and proinflammatory signaling pathways in SAE cells. In contrast, the effects of asbestos were minimal in ρ(0) SAE cells. CONCLUSIONS: Mitochondria are a major cytoplasmic target of asbestos. Asbestos may initiate mitochondria-associated ROS, which mediate asbestos-induced nuclear mutagenic events and inflammatory signaling pathways in exposed cells. These data provide new insights into the molecular mechanisms of asbestos-induced genotoxicity.


Subject(s)
Asbestos/adverse effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Analysis of Variance , DNA Damage , Deoxyguanosine/analogs & derivatives , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Micronucleus Tests , Microscopy, Fluorescence , Mitochondria/metabolism , Respiratory System/cytology , Reverse Transcriptase Polymerase Chain Reaction
19.
J Am Heart Assoc ; 1(6): e003855, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23316324

ABSTRACT

BACKGROUND: Mitochondrial dysfunction is a key determinant in chagasic cardiomyopathy development in mice; however, its relevance in human Chagas disease is not known. We determined if defects in mitochondrial biogenesis and dysregulation of peroxisome proliferator-activated receptor gamma (PPARγ) coactivator-1 (PGC-1)-regulated transcriptional pathways constitute a mechanism or mechanisms underlying mitochondrial oxidative-phosphorylation (OXPHOS) deficiency in human Chagas disease. METHODS AND RESULTS: We utilized human cardiomyocytes and left-ventricular tissue from chagasic and other cardiomyopathy patients and healthy donors (n>6/group). We noted no change in citrate synthase activity, yet mRNA and/or protein levels of subunits of the respiratory complexes were significantly decreased in Trypanosoma cruzi-infected cardiomyocytes (0 to 24 hours) and chagasic hearts. We observed increased mRNA and decreased nuclear localization of PGC-1-coactivated transcription factors, yet the expression of genes for PPARγ-regulated fatty acid oxidation and nuclear respiratory factor (NRF1/2)-regulated mtDNA replication and transcription machinery was enhanced in infected cardiomyocytes and chagasic hearts. The D-loop formation was normal or higher, but mtDNA replication and mtDNA content were decreased by 83% and 40% to 65%, respectively. Subsequently, we noted that reactive oxygen species (ROS), oxidative stress, and mtDNA oxidation were significantly increased, yet NRF1/2-regulated antioxidant gene expression remained compromised in infected cardiomyocytes and chagasic hearts. CONCLUSIONS: The replication of mtDNA was severely compromised, resulting in a significant loss of mtDNA and expression of OXPHOS genes in T cruzi-infected cardiomyocytes and chagasic hearts. Our data suggest increased ROS generation and selective functional incapacity of NRF2-mediated antioxidant gene expression played a role in the defects in mtDNA replication and unfitness of mtDNA for replication and gene expression in Chagas disease.


Subject(s)
Chagas Disease/physiopathology , DNA Replication/physiology , DNA, Mitochondrial/physiology , Mitochondrial Turnover/physiology , Trypanosoma cruzi , Blotting, Western , Cells, Cultured , Chagas Disease/genetics , Chagas Disease/metabolism , DNA, Mitochondrial/metabolism , Gene Expression Regulation , Humans , Immunohistochemistry , Microscopy, Fluorescence , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Myocytes, Cardiac/physiology , Myocytes, Cardiac/ultrastructure , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/physiology , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Nuclear Respiratory Factor 1/physiology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
20.
Int J Environ Res Public Health ; 8(6): 2090-108, 2011 06.
Article in English | MEDLINE | ID: mdl-21776218

ABSTRACT

Chronic arsenic exposure results in higher risk of skin, lung, and bladder cancer, as well as cardiovascular disease and diabetes. The purpose of this study was to investigate the effects on expression of selected genes in the blood lymphocytes from 159 people exposed chronically to arsenic in their drinking water using a novel RT-PCR TaqMan low-density array (TLDA). We found that expression of tumor necrosis factor-α (TNF-α), which activates both inflammation and NF-κB-dependent survival pathways, was strongly associated with water and urinary arsenic levels. Expression of KCNA5, which encodes a potassium ion channel protein, was positively associated with water and toe nail arsenic levels. Expression of 2 and 11 genes were positively associated with nail and urinary arsenic, respectively. Because arsenic exposure has been reported to be associated with long QT intervals and vascular disease in humans, we also used this TLDA for analysis of gene expression in human cardiomyocytes exposed to arsenic in vitro. Expression of the ion-channel genes CACNA1, KCNH2, KCNQ1 and KCNE1 were down-regulated by 1-µM arsenic. Alteration of some common pathways, including those involved in oxidative stress, inflammatory signaling, and ion-channel function, may underlay the seemingly disparate array of arsenic-associated diseases, such as cancer, cardiovascular disease, and diabetes.


Subject(s)
Arsenic/administration & dosage , Arsenic/metabolism , Gene Expression/drug effects , Myocytes, Cardiac/drug effects , Real-Time Polymerase Chain Reaction , Adolescent , Adult , Child , Dose-Response Relationship, Drug , Environmental Exposure , Female , Humans , Ion Channels/metabolism , Lymphocytes/drug effects , Male , Middle Aged , Myocytes, Cardiac/metabolism , Specimen Handling , Surveys and Questionnaires , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...